Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Nutr Biochem ; 111: 109153, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36150680

RESUMEN

This study aimed to characterize the potential beneficial effects of chronic docosahexaenoic acid (DHA) supplementation on restoring subcutaneous white adipose tissue (scWAT) plasticity in obese aged female mice. Two-month-old female C57BL/6J mice received a control (CT) or a high fat diet (HFD) for 4 months. Then, 6-month-old diet-induced obese (DIO) mice were distributed into the DIO and the DIOMEG group (fed with a DHA-enriched HFD) up to 18 months. In scWAT, the DHA-enriched diet reduced the mean adipocyte size and reversed the upregulation of lipogenic genes induced by the HFD, reaching values even lower than those observed in CT animals. DIO mice exhibited an up-regulation of lipolytic and fatty oxidation gene expressions that was reversed in DHA-supplemented mice except for Cpt1a mRNA levels, which were higher in DIOMEG as compared to CT mice. DHA restored the increase of proinflammatory genes observed in scWAT of DIO mice. While no changes were observed in total macrophage F4/80+/CD11b+ content, the DHA treatment switched scWAT macrophages profile by reducing the M1 marker Cd11c and increasing the M2 marker CD206. These events occurred alongside with a stimulation of beige adipocyte specific genes, the restoration of UCP1 and pAKT/AKT ratio, and a recovery of the HFD-induced Fgf21 upregulation. In summary, DHA supplementation induced a metabolic remodeling of scWAT to a healthier phenotype in aged obese mice by modulating genes controlling lipid accumulation in adipocytes, reducing the inflammatory status, and inducing beige adipocyte markers in obese aged mice.


Asunto(s)
Ácidos Docosahexaenoicos , Obesidad , Femenino , Ratones , Animales , Ratones Obesos , Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/metabolismo , Obesidad/metabolismo , Ratones Endogámicos C57BL , Tejido Adiposo Blanco/metabolismo , Dieta Alta en Grasa/efectos adversos , Grasa Subcutánea/metabolismo , Suplementos Dietéticos , Tejido Adiposo/metabolismo
2.
Nutrients ; 14(20)2022 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-36296923

RESUMEN

Obesity and aging promote chronic low-grade systemic inflammation. The aim of the study was to analyze the effects of long-term physical exercise and/or omega-3 fatty acid Docosahexaenoic acid (DHA) supplementation on genes or proteins related to muscle metabolism, inflammation, muscle damage/regeneration and myokine expression in aged and obese mice. Two-month-old C57BL/6J female mice received a control or a high-fat diet for 4 months. Then, the diet-induced obese (DIO) mice were distributed into four groups: DIO, DIO + DHA, DIO + EX (treadmill training) and DIO + DHA + EX up to 18 months. Mice fed a control diet were sacrificed at 2, 6 and 18 months. Aging increased the mRNA expression of Tnf-α and decreased the expression of genes related to glucose uptake (Glut1, Glut4), muscle atrophy (Murf1, Atrogin-1, Cas-9) and myokines (Metrnl, Il-6). In aged DIO mice, exercise restored several of these changes. It increased the expression of genes related to glucose uptake (Glut1, Glut4), fatty acid oxidation (Cpt1b, Acox), myokine expression (Fndc5, Il-6) and protein turnover, decreased Tnf-α expression and increased p-AKT/AKT ratio. No additional effects were observed when combining exercise and DHA. These data suggest the effectiveness of long-term training to prevent the deleterious effects of aging and obesity on muscle dysfunction.


Asunto(s)
Ácidos Docosahexaenoicos , Ácidos Grasos Omega-3 , Femenino , Ratones , Animales , Ratones Obesos , Ácidos Docosahexaenoicos/farmacología , Transportador de Glucosa de Tipo 1 , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratones Endogámicos C57BL , Dieta Alta en Grasa , Obesidad/metabolismo , Ácidos Grasos Omega-3/farmacología , Glucosa/metabolismo , Músculos/metabolismo , Envejecimiento , Inflamación , ARN Mensajero , Suplementos Dietéticos
3.
Nutrients ; 13(7)2021 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-34371972

RESUMEN

Resistance training (RT) and n-3 polyunsaturated fatty acids (n-3 PUFA) supplementation have emerged as strategies to improve muscle function in older adults. Overweight/obese postmenopausal women (55-70 years) were randomly allocated to one of four experimental groups, receiving placebo (olive oil) or docosahexaenoic acid (DHA)-rich n-3 PUFA supplementation alone or in combination with a supervised RT-program for 16 weeks. At baseline and at end of the trial, body composition, anthropometrical measures, blood pressure and serum glucose and lipid biomarkers were analyzed. Oral glucose tolerance tests (OGTT) and strength tests were also performed. All groups exhibit a similar moderate reduction in body weight and fat mass, but the RT-groups maintained bone mineral content, increased upper limbs lean mass, decreased lower limbs fat mass, and increased muscle strength and quality compared to untrained-groups. The RT-program also improved glucose tolerance (lowering the OGTT incremental area under the curve). The DHA-rich supplementation lowered diastolic blood pressure and circulating triglycerides and increased muscle quality in lower limbs. In conclusion, 16-week RT-program improved segmented body composition, bone mineral content, and glucose tolerance, while the DHA-rich supplement had beneficial effects on cardiovascular health markers in overweight/obese postmenopausal women. No synergistic effects were observed for DHA supplementation and RT-program combination.


Asunto(s)
Composición Corporal , Factores de Riesgo Cardiometabólico , Ácidos Docosahexaenoicos/administración & dosificación , Sobrepeso/terapia , Posmenopausia , Entrenamiento de Fuerza , Anciano , Glucemia/análisis , Suplementos Dietéticos , Método Doble Ciego , Ácidos Grasos Omega-3/administración & dosificación , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Metabolismo de los Lípidos , Persona de Mediana Edad , Fuerza Muscular , Obesidad/fisiopatología , Obesidad/terapia , Sobrepeso/fisiopatología , Placebos
4.
FASEB J ; 35(6): e21592, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33960028

RESUMEN

Brown adipose tissue (BAT) dysfunction in aging and obesity has been related to chronic unresolved inflammation, which could be mediated by an impaired production of specialized proresolving lipid mediators (SPMs), such as Lipoxins-LXs, Resolvins-Rvs, Protectins-PDs, and Maresins-MaRs. Our aim was to characterize the changes in BAT SPMs signatures and their association with BAT dysfunction during aging, especially under obesogenic conditions, and their modulation by a docosahexaenoic acid (DHA)-rich diet. Lipidomic, functional, and molecular studies were performed in BAT of 2- and 18-month-old lean (CT) female mice and in 18-month-old diet-induced obese (DIO) mice fed with a high-fat diet (HFD), or a DHA-enriched HFD. Aging downregulated Prdm16 and UCP1 levels, especially in DIO mice, while DHA partially restored them. Arachidonic acid (AA)-derived LXs and DHA-derived MaRs and PDs were the most abundant SPMs in BAT of young CT mice. Interestingly, the sum of LXs and of PDs were significantly lower in aged DIO mice compared to young CT mice. Some of the SPMs most significantly reduced in obese-aged mice included LXB4 , MaR2, 4S,14S-diHDHA, 10S,17S-diHDHA (a.k.a. PDX), and RvD6. In contrast, DHA increased DHA-derived SPMs, without modifying LXs. However, MicroPET studies showed that DHA was not able to counteract the impaired cold exposure response in BAT of obese-aged mice. Our data suggest that a defective SPMs production could underlie the decrease of BAT activity observed in obese-aged mice, and highlight the relevance to further characterize the physiological role and therapeutic potential of specific SPMs on BAT development and function.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Envejecimiento/patología , Suplementos Dietéticos , Ácidos Docosahexaenoicos/administración & dosificación , Lípidos/análisis , Obesidad/fisiopatología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/patología , Animales , Dieta Alta en Grasa , Femenino , Metabolismo de los Lípidos , Lipidómica , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Nutrients ; 13(2)2021 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-33546405

RESUMEN

Obesity and aging are associated to non-alcoholic fatty liver disease (NAFLD) development. Here, we investigate whether long-term feeding with a docosahexaenoic acid (DHA)-enriched diet and aerobic exercise, alone or in combination, are effective in ameliorating NAFLD in aged obese mice. Two-month-old female C57BL/6J mice received control or high fat diet (HFD) for 4 months. Then, the diet-induced obese (DIO) mice were distributed into four groups: DIO, DIO + DHA (15% dietary lipids replaced by a DHA-rich concentrate), DIO + EX (treadmill running), and DIO + DHA + EX up to 18 months. The DHA-rich diet reduced liver steatosis in DIO mice, decreasing lipogenic genes (Dgat2, Scd1, Srebp1c), and upregulated lipid catabolism genes (Hsl/Acox) expression. A similar pattern was observed in the DIO + EX group. The combination of DHA + exercise potentiated an increase in Cpt1a and Ppara genes, and AMPK activation, key regulators of fatty acid oxidation. Exercise, alone or in combination with DHA, significantly reversed the induction of proinflammatory genes (Mcp1, Il6, Tnfα, Tlr4) in DIO mice. DHA supplementation was effective in preventing the alterations induced by the HFD in endoplasmic reticulum stress-related genes (Ern1/Xbp1) and autophagy markers (LC3II/I ratio, p62, Atg7). In summary, long-term DHA supplementation and/or exercise could be helpful to delay NAFLD progression during aging in obesity.


Asunto(s)
Envejecimiento/fisiología , Ácidos Docosahexaenoicos/administración & dosificación , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Obesidad/complicaciones , Condicionamiento Físico Animal/fisiología , Animales , Autofagia/genética , Autofagia/fisiología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Inflamación/genética , Metabolismo de los Lípidos , Lipogénesis/genética , Hígado/química , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/etiología , ARN Mensajero/análisis
6.
Appl Physiol Nutr Metab ; 45(9): 957-967, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32176854

RESUMEN

Obesity is characterized by excessive fat accumulation and inflammation. Aging has also been characterized as an inflammatory condition, frequently accompanied by accumulation of visceral fat. Beneficial effects of exercise and n-3 long-chain polyunsaturated fatty acids in metabolic disorders have been described. Glucose transporter 12 (GLUT12) is one of the less investigated members of the GLUT family. Glucose, insulin, and tumor necrosis factor alpha (TNF-α) induce GLUT12 translocation to the membrane in muscle, adipose tissue, and intestine. We aimed to investigate GLUT12 expression in obesity and aging, and under diet supplementation with docosahexaenoic acid (DHA) alone or in combination with physical exercise in mice. Aging increased GLUT12 expression in intestine, kidney, and adipose tissue, whereas obesity reduced it. No changes on the transporter occurred in skeletal muscle. In obese 18-month-old mice, DHA further decreased GLUT12 in the 4 organs. Aerobic exercise alone did not modify GLUT12, but the changes triggered by exercise were able to prevent the DHA-diminishing effect, and almost restored GLUT12 basal levels. In conclusion, the downregulation of metabolism in aging would be a stimulus to upregulate GLUT12 expression. Contrary, obesity, an excessive energy condition, would induce GLUT12 downregulation. The combination of exercise and DHA would contribute to restore basal function of GLUT12. Novelty In small intestine, kidney and adipose tissue aging increases GLUT12 protein expression whereas obesity reduces it. Dietary DHA decreases GLUT12 in small intestine, kidney, adipose tissue and skeletal muscle. Exercise alone does not modify GLUT12 expression, nevertheless exercise prevents the DHA-diminishing effect on GLUT12.


Asunto(s)
Envejecimiento/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Obesidad/metabolismo , Condicionamiento Físico Animal , Tejido Adiposo/metabolismo , Animales , Células CACO-2 , Dieta , Femenino , Humanos , Intestino Delgado/metabolismo , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo
7.
J Physiol Biochem ; 76(2): 251-267, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31853728

RESUMEN

Adipose tissue dysfunction represents the hallmark of obesity. Brown/beige adipose tissues play a crucial role in maintaining energy homeostasis through non-shivering thermogenesis. Brown adipose tissue (BAT) activity has been inversely related to body fatness, suggesting that BAT activation is protective against obesity. BAT plays also a key role in the control of triglyceride clearance, glucose homeostasis, and insulin sensitivity. Therefore, BAT/beige activation has been proposed as a strategy to prevent or ameliorate obesity development and associated commorbidities. In the last few years, a variety of preclinical studies have proposed n-3 polyunsaturated fatty acids (n-3 PUFAs) as novel inducers of BAT activity and white adipose tissue browning. Here, we review the in vitro and in vivo available evidences of the thermogenic properties of n-3 PUFAs, especially focusing on the molecular and cellular physiological mechanisms involved. Finally, we also discuss the challenges and future perspectives to better characterize the therapeutic potential of n-3 PUFAs as browning agents, especially in humans.


Asunto(s)
Tejido Adiposo Beige/metabolismo , Tejido Adiposo Pardo/metabolismo , Ácidos Grasos Omega-3 , Obesidad , Animales , Células Cultivadas , Metabolismo Energético , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-3/fisiología , Humanos , Ratones , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Ratas , Termogénesis
8.
J Nutr Biochem ; 76: 108264, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31760230

RESUMEN

Tumor necrosis factor-alfa (TNF-α) is a pro-inflammatory cytokine highly-involved in intestinal inflammation. Omega-3 polyunsaturated fatty acids (n3-PUFAs) show anti-inflammatory actions. We previously demonstrated that the n3-PUFA EPA prevents TNF-α inhibition of sugar uptake in Caco-2 cells. Here, we investigated whether the n3-PUFA DHA and its derived specialized pro-resolving lipid mediators (SPMs) MaR1, RvD1 and RvD2, could block TNF-α inhibition of intestinal sugar and glutamine uptake. DHA blocked TNF-α-induced inhibition of α-methyl-D-glucose (αMG) uptake and SGLT1 expression in the apical membrane of Caco-2 cells, through a pathway independent of GPR120. SPMs showed the same preventive effect but acting at concentrations 1000 times lower. In diet-induced obese (DIO) mice, oral gavage of MaR1 reversed the up-regulation of pro-inflammatory cytokines found in intestinal mucosa of these mice. However, MaR1 treatment was not able to counteract the reduced intestinal transport of αMG and SGLT1 expression in the DIO mice. In Caco-2 cells, TNF-α also inhibited glutamine uptake being this inhibition prevented by EPA, DHA and the DHA-derived SPMs. Interestingly, TNF-α increased the expression in the apical membrane of the glutamine transporter B0AT1. This increase was partially blocked by the n-3 PUFAs. These data reveal DHA and its SPMs as promising biomolecules to restore intestinal nutrients transport during intestinal inflammation.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Glutamina/metabolismo , Lípidos/química , Azúcares/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Transporte Biológico/efectos de los fármacos , Biotinilación , Células CACO-2 , Dieta , Ácido Eicosapentaenoico/farmacología , Ácidos Grasos Omega-3/farmacología , Humanos , Inflamación , Mucosa Intestinal/metabolismo , Intestinos/química , Intestinos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Transportador 1 de Sodio-Glucosa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Nutrients ; 11(10)2019 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-31635188

RESUMEN

Type 2 diabetes (T2D) is a complex metabolic disease, which involves a maintained hyperglycemia due to the development of an insulin resistance process. Among multiple risk factors, host intestinal microbiota has received increasing attention in T2D etiology and progression. In the present study, we have explored the effect of long-term supplementation with a non-dairy fermented food product (FFP) in Zucker Diabetic and Fatty (ZDF) rats T2D model. The supplementation with FFP induced an improvement in glucose homeostasis according to the results obtained from fasting blood glucose levels, glucose tolerance test, and pancreatic function. Importantly, a significantly reduced intestinal glucose absorption was found in the FFP-treated rats. Supplemented animals also showed a greater survival suggesting a better health status as a result of the FFP intake. Some dissimilarities have been observed in the gut microbiota population between control and FFP-treated rats, and interestingly a tendency for better cardiometabolic markers values was appreciated in this group. However, no significant differences were observed in body weight, body composition, or food intake between groups. These findings suggest that FFP induced gut microbiota modifications in ZDF rats that improved glucose metabolism and protected from T2D development.


Asunto(s)
Diabetes Mellitus Tipo 2/prevención & control , Alimentos Fermentados , Lactobacillales , Animales , Glucemia , Composición Corporal , Peso Corporal , Suplementos Dietéticos , Fermentación , Alimentos Funcionales , Intolerancia a la Glucosa , Masculino , Ratas , Ratas Zucker
10.
Nutrients ; 11(4)2019 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-31003450

RESUMEN

Aging is a complex phenomenon characterized by the progressive loss of tissue and organ function. The oxidative-stress theory of aging postulates that age-associated functional losses are due to the accumulation of ROS-induced damage. Liver function impairment and non-alcoholic fatty liver disease (NAFLD) are common among the elderly. NAFLD can progress to non-alcoholic steatohepatitis (NASH) and evolve to hepatic cirrhosis or hepatic carcinoma. Oxidative stress, lipotoxicity, and inflammation play a key role in the progression of NAFLD. A growing body of evidence supports the therapeutic potential of omega-3 polyunsaturated fatty acids (n-3 PUFA), mainly docosahaexenoic (DHA) and eicosapentaenoic acid (EPA), on metabolic diseases based on their antioxidant and anti-inflammatory properties. Here, we performed a systematic review of clinical trials analyzing the efficacy of n-3 PUFA on both systemic oxidative stress and on NAFLD/NASH features in adults. As a matter of fact, it remains controversial whether n-3 PUFA are effective to counteract oxidative stress. On the other hand, data suggest that n-3 PUFA supplementation may be effective in the early stages of NAFLD, but not in patients with more severe NAFLD or NASH. Future perspectives and relevant aspects that should be considered when planning new randomized controlled trials are also discussed.


Asunto(s)
Ácidos Grasos Omega-3/farmacología , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/etiología , Estrés Oxidativo/efectos de los fármacos , Envejecimiento , Humanos
11.
Lipids Health Dis ; 17(1): 103, 2018 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-29743087

RESUMEN

BACKGROUND: Eicosapentaenoic acid (EPA) and α-lipoic acid (α-LA) have been investigated for their beneficial effects on obesity and cardiovascular risk factors. In the current research, the goal was to evaluate metabolomic changes following the dietary supplementation of these two lipids, alone or combined in healthy overweight/obese sedentary women following an energy-restricted diet. For this purpose, an untargeted metabolomics approach was conducted on urine samples using liquid chromatography coupled with time of flight mass spectrometry (HPLC-TOF-MS). METHODS: This is a short-term double blind placebo-controlled study with a parallel nutritional design that lasted 10 weeks. Participants were assigned to one of the 4 experimental groups [Control, EPA (1.3 g/d), α-LA (0.3 g/d) and EPA+α-LA (1.3 g/d + 0.3 g/d)]. All intervention groups followed an energy-restricted diet of 30% less than total energy expenditure. Clinically relevant biochemical measurements were analyzed. Urine samples (24 h) were collected at baseline and after 10 weeks. Untargeted metabolomic analysis on urine samples was carried out, and principal component analysis (PCA) and partial least squares-discriminant analysis (PLS-DA) were performed for the pattern recognition and characteristic metabolites identification. RESULTS: Urine samples were scattered in the PCA scores plots in response to the supplementation with α-LA. Totally, 28 putative discriminant metabolites in positive ionization, and 6 in negative ionization were identified among groups clearly differentiated according to the α-LA administration. Remarkably is the presence of an ascorbate intermediate metabolite (one of the isomers of trihydroxy-dioxohexanoate, or dihydroxy-oxohexanedionate) in the groups supplemented with α-LA. This fact might be associated with antioxidant properties of both α-LA and ascorbic acid. Correlations between phenotypical parameters and putative metabolites of provided additional information on whether there is a direct or inverse relationship between them. Especially interesting are the negative correlation between ascorbate intermediate metabolite and asymmetric dimethylarginine (ADMA) and the positive one between superoxide dismutase (SOD) and α-LA supplementation. CONCLUSIONS: This metabolomic approach supports that the beneficial effects of α-LA administration on body weight reduction may be partly explained by the antioxidant properties of this organosulfur carboxylic acid mediated by isomers of trihydroxy-dioxohexanoate, or dihydroxy-oxohexanedionate. TRIAL REGISTRATION: Clinicaltrials.gov NCT01138774 .


Asunto(s)
Ácido Eicosapentaenoico/administración & dosificación , Obesidad/dietoterapia , Sobrepeso/dietoterapia , Ácido Tióctico/administración & dosificación , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/fisiopatología , Adulto , Antioxidantes/metabolismo , Enfermedades Cardiovasculares/dietoterapia , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/prevención & control , Suplementos Dietéticos , Femenino , Voluntarios Sanos , Humanos , Masculino , Metabolómica/métodos , Obesidad/fisiopatología , Sobrepeso/fisiopatología , Factores de Riesgo , Pérdida de Peso/efectos de los fármacos
12.
Biofactors ; 43(1): 117-131, 2017 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-27507611

RESUMEN

In obesity, the increment of adiposity levels disrupts the whole body homeostasis, promoting an over production of oxidants and inflammatory mediators. The current study aimed to characterize the transcriptomic changes promoted by supplementation with eicosapentaenoic acid (EPA, 1.3 g/day), α-lipoic acid (0.3 g/day), or both (EPA + α-lipoic acid, 1.3 g/day + 0.3 g/day) in subcutaneous abdominal adipose tissue from overweight/obese healthy women, who followed a hypocaloric diet (30% of total energy expenditure) during ten weeks, by using a microarray approach. At the end of the intervention, a total of 33,297 genes were analyzed using Affymetrix GeneChip arrays. EPA promoted changes in extracellular matrix remodeling gene expression, besides a rise of genes associated with either chemotaxis or wound repair. α-Lipoic acid decreased expression of genes related with cell adhesion and inflammation. Furthermore, α-lipoic acid, especially in combination with EPA, upregulated the expression of genes associated with lipid catabolism while downregulated genes involved in lipids storage. Together, all these data suggest that some of the metabolic effects of EPA and α-lipoic acid could be related to their regulatory actions on adipose tissue metabolism. © 2016 BioFactors, 43(1):117-131, 2017.


Asunto(s)
Antiinflamatorios/administración & dosificación , Restricción Calórica , Ácido Eicosapentaenoico/administración & dosificación , Obesidad/dietoterapia , Ácido Tióctico/administración & dosificación , Transcriptoma , Grasa Abdominal/metabolismo , Administración Oral , Adulto , Suplementos Dietéticos , Femenino , Humanos , Redes y Vías Metabólicas , Persona de Mediana Edad , Obesidad/metabolismo
13.
Prostaglandins Other Lipid Mediat ; 121(Pt A): 24-41, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26219838

RESUMEN

The n-3 long-chain polyunsaturated fatty acids (n-3 PUFAs) such as eicosapentaenoic (EPA) and docosahexaenoic (DHA) have been reported to improve obesity-associated metabolic disorders including chronic inflammation, insulin resistance and dyslipidaemia. Growing evidence exits about adipose tissue as a target in mediating the beneficial effects of these marine n-3 PUFAs in adverse metabolic syndrome manifestations. Therefore, in this manuscript we focus in reviewing the current knowledge about effects of marine n-3 PUFAs on adipose tissue metabolism and secretory functions. This scope includes n-3 PUFAs actions on adipogenesis, lipogenesis and lipolysis as well as on fatty acid oxidation and mitochondrial biogenesis. The effects of n-3 PUFAs on adipose tissue glucose uptake and insulin signaling are also summarized. Moreover, the roles of peroxisome proliferator-activated receptor γ (PPARγ) and AMPK activation in mediating n-3 PUFAs actions on adipose tissue functions are discussed. Finally, the mechanisms underlying the ability of n-3 PUFAs to prevent and/or ameliorate adipose tissue inflammation are also revised, focusing on the role of n-3 PUFAs-derived specialized proresolving lipid mediators such as resolvins, protectins and maresins.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Ácidos Grasos Omega-3/farmacología , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/patología , Obesidad/tratamiento farmacológico , Obesidad/patología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Ácidos Grasos Omega-3/uso terapéutico , Humanos , Síndrome Metabólico/metabolismo , Obesidad/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo
14.
J. physiol. biochem ; 71(2): 341-349, jun. 2015.
Artículo en Inglés | IBECS | ID: ibc-140541

RESUMEN

Inflammation is involved in the pathophysiology of many chronic diseases, such as rheumatoid arthritis and neurodegenerative diseases. Several studies have evidenced important anti-inflammatory and immunomodulatory properties of omega-3 long-chain polyunsaturated fatty acids (n-3 LC-PUFAs). This review illustrates current knowledge about the efficacy of n-3 LC-PUFAs (eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), particularly) in preventing and/or treating several chronic inflammatory conditions (inflammatory bowel diseases and rheumatoid arthritis) as well as their potential benefits on neurodegenerative diseases. It is well established that n-3 LC-PUFAs are substrates for synthesis of novel series of lipid mediators (e.g., resolvins, protectins, and maresins) with potent anti-inflammatory and pro-resolving properties, which have been proposed to partly mediate the protective and beneficial actions of n-3 LC-PUFAs. Here, we briefly summarize current knowledge from preclinical studies analyzing the actions of EPA- and DHA-derived resolvins and protectins on pathophysiological models of rheumatoid arthritis, Alzheimer, and irritable bowel syndrome


Asunto(s)
Humanos , Enfermedades Neurodegenerativas/fisiopatología , Artritis Reumatoide/fisiopatología , Ácidos Grasos Omega-3/farmacocinética , Sustancias Protectoras/farmacocinética , Ácido Eicosapentaenoico/farmacocinética
15.
J Physiol Biochem ; 71(2): 341-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25752887

RESUMEN

Inflammation is involved in the pathophysiology of many chronic diseases, such as rheumatoid arthritis and neurodegenerative diseases. Several studies have evidenced important anti-inflammatory and immunomodulatory properties of omega-3 long-chain polyunsaturated fatty acids (n-3 LC-PUFAs). This review illustrates current knowledge about the efficacy of n-3 LC-PUFAs (eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), particularly) in preventing and/or treating several chronic inflammatory conditions (inflammatory bowel diseases and rheumatoid arthritis) as well as their potential benefits on neurodegenerative diseases. It is well established that n-3 LC-PUFAs are substrates for synthesis of novel series of lipid mediators (e.g., resolvins, protectins, and maresins) with potent anti-inflammatory and pro-resolving properties, which have been proposed to partly mediate the protective and beneficial actions of n-3 LC-PUFAs. Here, we briefly summarize current knowledge from preclinical studies analyzing the actions of EPA- and DHA-derived resolvins and protectins on pathophysiological models of rheumatoid arthritis, Alzheimer, and irritable bowel syndrome.


Asunto(s)
Ácidos Grasos Omega-3/farmacología , Inflamación/prevención & control , Enfermedades Neurodegenerativas/prevención & control , Animales , Artritis Reumatoide/fisiopatología , Artritis Reumatoide/prevención & control , Antígenos CD59/metabolismo , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Ácidos Grasos Omega-3/inmunología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/prevención & control , Enfermedades Neurodegenerativas/metabolismo
16.
Expert Opin Ther Targets ; 19(7): 893-909, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25726860

RESUMEN

INTRODUCTION: Leptin is a hormone with a key role in food intake and body weight homeostasis. Congenital leptin deficiency (CLD) is a rare disease that causes hyperphagia and early severe obesity. However, common obesity conditions are associated with hyperleptinemia and leptin resistance. AREAS COVERED: The main signaling pathways activated by leptin as well as the mechanisms underlying the regulatory actions of leptin on food intake and on lipid and glucose metabolism are reviewed. The potential mechanisms involving leptin resistance and the main regulatory hormonal and nutritional factors controlling leptin production/functions are also analyzed. The pathophysiology of leptin in human obesity, and especially the trials analyzing effects of leptin replacement therapy in patients with CLD or in subjects with common obesity and in post-obese weight-reduced subjects are also summarized. EXPERT OPINION: The use of drugs or specific bioactive food components with anti-inflammatory properties to reduce the inflammatory state associated with obesity, especially at the hypothalamus, may help to overcome leptin resistance. Research should also be focused on investigating dietary strategies, food supplements or drugs capable of avoiding or reversing the leptin fall during weight management, in order to promote sustained body weight lowering and weight loss maintenance.


Asunto(s)
Fármacos Antiobesidad/farmacología , Leptina/metabolismo , Obesidad/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Peso Corporal , Humanos , Hipotálamo/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/fisiopatología , Leptina/deficiencia , Terapia Molecular Dirigida , Obesidad/fisiopatología , Transducción de Señal/efectos de los fármacos
17.
Obesity (Silver Spring) ; 23(2): 313-21, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25594166

RESUMEN

OBJECTIVE: To evaluate the potential body weight-lowering effects of dietary supplementation with eicosapentaenoic acid (EPA) and α-lipoic acid separately or combined in healthy overweight/obese women following a hypocaloric diet. METHODS: This is a short-term double-blind placebo-controlled study with parallel design that lasted 10 weeks. Of the randomized participants, 97 women received the allocated treatment [Control, EPA (1.3 g/d), α-lipoic acid (0.3 g/d), and EPA+α-lipoic acid (1.3 g/d+0.3 g/d)], and 77 volunteers completed the study. All groups followed an energy-restricted diet of 30% less than total energy expenditure. Body weight, anthropometric measurements, body composition, resting energy expenditure, blood pressure, serum glucose, and insulin and lipid profile, as well as leptin and ghrelin levels, were assessed at baseline and after nutritional intervention. RESULTS: Body weight loss was significantly higher (P<0.05) in those groups supplemented with α-lipoic acid. EPA supplementation significantly attenuated (P<0.001) the decrease in leptin levels that occurs during weight loss. Body weight loss improved lipid and glucose metabolism parameters but without significant differences between groups. CONCLUSIONS: The intervention suggests that α-lipoic acid supplementation alone or in combination with EPA may help to promote body weight loss in healthy overweight/obese women following energy-restricted diets.


Asunto(s)
Antioxidantes/administración & dosificación , Ácido Eicosapentaenoico/administración & dosificación , Obesidad/tratamiento farmacológico , Ácido Tióctico/administración & dosificación , Pérdida de Peso , Administración Oral , Adulto , Composición Corporal , Colesterol/sangre , Suplementos Dietéticos , Método Doble Ciego , Quimioterapia Combinada , Ácido Eicosapentaenoico/sangre , Metabolismo Energético , Femenino , Ghrelina/metabolismo , Glucosa/metabolismo , Humanos , Insulina/sangre , Leptina/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Persona de Mediana Edad , Obesidad/sangre , Ácido Tióctico/sangre , Resultado del Tratamiento , Adulto Joven
18.
J Nutr ; 146(4): 889S-896S, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26962183

RESUMEN

BACKGROUND: The proinflammatory state induced by obesity plays an important role in obesity-related metabolic complications. OBJECTIVE: Our objective was to evaluate whether dietary supplementation with α-lipoic acid (LA) and eicosapentaenoic acid (EPA), separately or in combination, could improve inflammatory and cardiovascular disease risk markers in healthy overweight or obese women consuming an energy-restricted diet. METHODS: Within the context of the Effects of Lipoic Acid and Eicosapentaenoic Acid in Human Obesity (OBEPALIP) study, Caucasian women (n = 73) aged 20-50 y with a BMI (in kg/m2) between 27.5 and 40 consumed an energy-restricted diet for 10 wk after being randomly assigned to 1 of 4 parallel experimental groups: a control group or groups supplemented with 1.3 g EPA/d, 0.3 g LA/d, or both. Secondary outcomes were measured at baseline and at the end of the study. These included circulating inflammatory [C-reactive protein (CRP), adiponectin, interleukin 6 (IL-6), chemerin, haptoglobin, amyloid A, and leukocytes] and cardiovascular disease risk markers (platelet count and circulating apelin, asymmetric dimethylarginine, vascular endothelial growth factor, and plasminogen activator inhibitor 1). Gene expression of IL6, adhesion G protein-coupled receptor E1 (ADGRE1), interleukin 10 (IL10), chemokine (C-C motif) ligand 2, and adiponectin was measured in subcutaneous abdominal adipose tissue biopsies at endpoint. RESULTS: Supplementation with LA caused a greater reduction in some circulating inflammatory risk markers, such as CRP (-0.13 ± 0.07 mg/dL compared with 0.06 ± 0.07 mg/dL, P < 0.05) and leukocyte count (-0.74 ± 0.18 × 103/mm3 compared with 0.06 ± 0.18 × 103/mm3, P < 0.01), than in the groups that were not supplemented with LA. In contrast, the fall in apelin concentrations that accompanied weight loss was less pronounced in groups that were supplemented with LA (-1.1 ± 4.9 pg/mL) than in those that were not (-21.3 ± 4.8 pg/mL, P < 0.01). In adipose tissue, compared with those who did not receive EPA, EPA-supplemented groups exhibited a downregulation of ADGRE1 (0.7 ± 0.1-fold compared with 1.0 ± 0.1-fold) (P < 0.05) and an upregulation of IL10 (1.8 ± 0.2-fold compared with 1.0 ± 0.2-fold) (P < 0.05) gene expression. CONCLUSIONS: Dietary supplementation with LA improves some systemic inflammatory and cardiovascular disease-related risk markers in healthy overweight or obese women independently of weight loss, whereas EPA modulates inflammation-related genes in adipose tissue. This trial was registered at clinicaltrials.gov as NCT01138774.

19.
J. physiol. biochem ; 69(3): 595-600, sept. 2013.
Artículo en Inglés | IBECS | ID: ibc-121678

RESUMEN

Lipoic acid (LA) is a naturally occurring compound with antioxidant properties. Recent attention has been focused on the potential beneficial effects of LA on obesity and related metabolic disorders. Dietary supplementation with LA prevents insulin resistance and upregulates adiponectin, an insulin-sensitizing adipokine, in obese rodents. The aim of this study was to investigate the direct effects of LA on adiponectin production in cultured adipocytes, as well as the potential signaling pathways involved. For this purpose, fully differentiated 3T3-L1 adipocytes were treated with LA (1-500 μM) during 24 h. The amount of adiponectin secreted to media was detected by ELISA, while adiponectin mRNA expression was determined by RT-PCR. Treatment with LA induced a dose-dependent inhibition on adiponectin gene expression and protein secretion. Pretreatment with the PI3K inhibitor LY294002 inhibited adiponectin secretion and mRNA levels, and significantly potentiated the inhibitory effect of LA on adiponectin secretion. The AMPK activator AICAR also reduced adiponectin production, but surprisingly, it was able to reverse the LA-induced inhibition of adiponectin. The JNK inhibitor SP600125 and the MAPK inhibitor PD98059 did not modify the inhibitory effect of LA on adiponectin. In conclusion, our results revealed that LA reduces adiponectin secretion in 3T3-L1 adipocytes, which contrasts with the stimulation of adiponectin described after in vivo supplementation with LA, suggesting that an indirect mechanism or some in vivo metabolic processing is involved


Asunto(s)
Humanos , Ácido Tióctico/farmacocinética , Adiponectina , Adipocitos , Obesidad/prevención & control , Sustancias Protectoras/farmacocinética , Antioxidantes/farmacocinética
20.
J. physiol. biochem ; 69(3): 633-651, sept. 2013.
Artículo en Inglés | IBECS | ID: ibc-121682

RESUMEN

The present review aims to illustrate current knowledge about the efficacy of omega-3 long-chain polyunsaturated fatty acids (n−3 LC-PUFAs) in treating/preventing several metabolic pathologies. We reviewed systematically the published evidence on the effectiveness of n−3 LC-PUFAs fish consumption or n−3 LC-PUFAs supplementation on prevention/treatment of obesity, metabolic syndrome, and cardiovascular diseases. Most of the reviewed studies were randomized-controlled interventional trials, although some relevant prospective and cross-sectional studies as well as some meta-analysis were also reviewed. Supplementation with n−3 LC-PUFAs might improve some obesity-associated metabolic syndrome features such as insulin resistance, hypertension and dyslipidemia by decreasing plasma triglycerides. Moreover, the blood pressure-lowering and anti-inflammatory properties of these fatty acids and their benefits in vascular function might confer cardioprotection. However, the efficacy of n−3 LC-PUFA on reducing myocardial infarction, arrhythmia, cardiac and sudden death, or stroke is controversial. Due to the beneficial actions of n−3 LC-PUFAs, several worldwide government and health organizations have established some recommendations of n−3 LC-PUFAs intake for groups of population. In general, the recommended levels for diseases prevention are lower than those advised for particular treatments. However, more clinical trials are necessary to recommend the most effective dosages and formulas (type of n−3 LC-PUFA, EPA/DHA ratio) for specific pathologies (AU)


Asunto(s)
Humanos , Ácidos Grasos Omega-3/farmacocinética , Obesidad/fisiopatología , Síndrome Metabólico/fisiopatología , Enfermedades Cardiovasculares/fisiopatología , Sustancias Protectoras/farmacocinética , Práctica Clínica Basada en la Evidencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA